Next Article in Journal
Heterologously Expressed Cellobiose Dehydrogenase Acts as Efficient Electron-Donor of Lytic Polysaccharide Monooxygenase for Cellulose Degradation in Trichoderma reesei
Next Article in Special Issue
Performance of a Shotgun Prediction Model for Colorectal Cancer When Using 16S rRNA Sequencing Data
Previous Article in Journal
A Randomized, Open-Label, Single-Dose, Crossover Study of the Comparative Bioavailability of EPA and DHA in a Novel Liquid Crystalline Nanoparticle-Based Formulation of ω-3 Acid Ethyl Ester Versus Omacor® Soft Capsule among Healthy Adults
Previous Article in Special Issue
How Can the Microbiome Induce Carcinogenesis and Modulate Drug Resistance in Cancer Therapy?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Role of the Gut and Intratumoral Microbiomes in Tumor Progression and Metastasis

by
Aneta Sevcikova
1,
Beata Mladosievicova
2,
Michal Mego
3 and
Sona Ciernikova
1,*
1
Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
2
Institute of Pathological Physiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
3
2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(24), 17199; https://doi.org/10.3390/ijms242417199
Submission received: 31 October 2023 / Revised: 30 November 2023 / Accepted: 4 December 2023 / Published: 6 December 2023
(This article belongs to the Special Issue Microbiota and Cancer 3.0)

Abstract

:
Cancer cell dissemination involves invasion, migration, resistance to stressors in the circulation, extravasation, colonization, and other functions responsible for macroscopic metastases. By enhancing invasiveness, motility, and intravasation, the epithelial-to-mesenchymal transition (EMT) process promotes the generation of circulating tumor cells and their collective migration. Preclinical and clinical studies have documented intensive crosstalk between the gut microbiome, host organism, and immune system. According to the findings, polymorphic microbes might play diverse roles in tumorigenesis, cancer progression, and therapy response. Microbial imbalances and changes in the levels of bacterial metabolites and toxins promote cancer progression via EMT and angiogenesis. In contrast, a favorable microbial composition, together with microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), can attenuate the processes of tumor initiation, disease progression, and the formation of distant metastases. In this review, we highlight the role of the intratumoral and gut microbiomes in cancer cell invasion, migration, and metastatic ability and outline the potential options for microbiota modulation. As shown in murine models, probiotics inhibited tumor development, reduced tumor volume, and suppressed angiogenesis and metastasis. Moreover, modulation of an unfavorable microbiome might improve efficacy and reduce treatment-related toxicities, bringing clinical benefit to patients with metastatic cancer.

Graphical Abstract

1. Introduction

The emerging trend of microbiome research in oncology results from studies uncovering the role of microorganisms in the etiology of several malignancies. Preclinical and clinical studies have also revealed a significant impact of the gut and tumor microbiomes on the efficacy of antitumor therapy and treatment-induced toxicity [1]. Moreover, mounting research focuses on the analysis of the microbiome composition in metastatic disease [2]. The significant role of the microbiome in oncogenesis and treatment underlines the fact that polymorphic microbiomes, including intestinal, oral, skin, tumor, lung, and vaginal microbiomes, were added to the extended comprehensive concept termed “The Hallmarks of Cancer”, which summarizes the key characteristics of tumors. The microbiome directly interacts positively or negatively with other hallmarks of malignancies, such as inflammation, immune impairment, genomic instability, and resistance to antitumor therapy [3].
The study of metastasis biology at the cellular, molecular, biochemical, and physical levels has undergone dramatic growth over the last 20 years. While the precise pathways are still under investigation, recent research has indicated new roles of cancer cells, which involve promoting genes with metastasis-driving mutations, cancer stem cells, circulating tumor cells (CTCs), epithelial-to-mesenchymal transition (EMT), and the metastatic dormancy and dynamic plasticity of cancer cells [4,5]. Various studies also demonstrated that the following drive metastatic spread: systemic inflammation; immune system modulation; specific interactions between cancer cells, immune cells, and cells in the tumor microenvironment; the avoidance of anoikis; immune checkpoint regulation; self-seeding, and other mechanisms. Mounting research highlights the role of the intratumoral and mucosal microbiomes in the progression of metastatic processes.
The mechanisms by which the intratumoral microbiota is implicated in metastatic spread are still unclear [6]. Recently, a newly developed single-cell RNA-sequencing method, known as INVADEseq, aimed to identify cell-associated bacteria in patient tumor samples and describe the pathways involved in metastatic processes [7]. According to the findings, the highly organized distribution of bacteria within tumors affects immune and epithelial cell functions, leading to disease progression [7]. Studies reported that specific microorganisms might affect metastatic processes, including EMT, resistance to fluid shear stress, immune system modulation, and matrix metalloproteinase (MMP) regulation [8]. EMT-related pathways might be regulated by specific microbial pathogens [9,10]. Moreover, the microbiome can change the actin cytoskeleton, contributing to tumor cells’ resistance to fluid shear stress [11]. As previously shown, Helicobacter pylori supports the remodulation of actin filaments, which results in EMT onset [12,13]. In addition, specific microbes within the microbiome contribute to tumor cell adaptation to specific biochemical factors in the tumor microenvironment during metastatic processes [8].
In this review, we aim to summarize the current knowledge about the emerging role of the gut and intratumoral microbiomes in metastasis and cancer progression. We analyze studies focusing on the association between the composition of microbial communities and metastatic disease in patients with different types of malignancies. Moreover, we outline the mechanisms of microbiome involvement in cancer progression and metastasis-related processes, including EMT and angiogenesis. Microbiota modulation represents an evolving phenomenon in cancer patient care, and mounting evidence supports the clinical utility of a microbiome-based approach in cancer initiation, progression, and treatment.

2. The Mechanisms of Tumor Progression and Metastasis

Tumor progression and metastasis represent multi-step processes, resulting in cancer cell changes that enable them to grow, spread, and establish secondary tumors at distant body sites (Figure 1).
The activation of invasion and metastasis is initiated by epigenetic changes, cell–cell interactions, growth factors, cytokines, signals from extracellular matrix components, extracellular matrix mechanical pressures, and the intratumoral microbiota [14].
The metastatic cascade includes the detachment of cancer cells from the primary tumor and the gaining of an invasive phenotype, local invasion into surrounding tissue, intravasation into the circulation, systemic transportation, extravasation, and the formation of colonies at distant sites, with adaptation and proliferation in secondary organs.
CTCs typically arise from epithelial tumor cells that undergo EMT, resulting in the loss of cell–cell adhesion and apical–basal polarity, the reorganization of the cytoskeleton, acquiring properties of tumor stem cells, and resistance to therapy. This process is regulated by transcription factors in tumor cells (Snail 1, Slug, ZEB1, Twist, FOXC2, etc.) and signaling pathways from the tumor microenvironment (WNT, Notch, Hedgehog, TGFβ, FGF, EGF, HGF signaling, etc.). Additionally, the hypoxia and activation of specific signaling pathways, including PI3K, WNT/β-catenin, and MAPK, affect EMT regulation [15,16]. Many studies focus not only on CTC detection and enumeration but also on CTC biomarkers, among which EMT markers are of great interest [17,18,19]. The most aggressive CTCs are related to the infiltration of the primary tumor or established metastasis in a process of “self-seeding”. Self-seeding in metastasis is the recruitment of cancer cells and the re-seeding of primary tumors and existing metastases by aggressive cancer cell clones [20,21].
Cancer cells can induce neutrophils to release neutrophil extracellular traps (NETs), which sequester CTCs and promote the metastatic process [22,23,24,25,26]. A certain number of CTCs can be eliminated by anoikis, the programmed apoptosis of cells [27]. However, cells can develop an anoikis-resistant state via oncogene activation (e.g., ERBB2 and RAS), an integrin switch (e.g., the downregulation of αvβ3 integrin expression), the constitutive activation of antiapoptotic pathways (e.g., the PI3K/Akt signaling pathway), the triggering of EMT, microRNAs (e.g., the downregulation of the miR200 family), high oxidative stress (e.g., activated growth factor receptors increase intracellular reactive oxygen species production by activating enzymes such as NADPH oxidase and lipoxygenase), hypoxia, the modulation of extracellular matrix stiffness, and the metabolic reprogramming of cancer cells [28]. Tumor cells can attach to specific distant organs/tissues and form colonies through distinct adhesion molecules, including proteoglycans (e.g., CD44), mucins (e.g., MUC16), integrins (e.g., α2β1), and the members of the immunoglobulin superfamily (e.g., ICAM1, VCAM1, and L1CAM) [29].
Before the arrival of tumor cells from primary tumors to the premetastatic niche [30], hematopoietic progenitor cells (VEGFR1-positive) travel from the bone marrow into the circulation and establish themselves in secondary organs, where they adhere to fibronectin, produced by fibroblasts and fibroblast-like cells [31]. The adherence is mediated by the integrin VLA-4, expressed by hematopoietic progenitor cells [32]. The nidation of tumor cells is primarily influenced by stromal-derived factor 1 (SDF-1), binding to the chemokine receptor CXCR4 [33]. CXCR4 receptor expression on breast cancer tumor cells is a typical determinant of bone metastasis [34,35]. Its activation results in pseudopodia formation and integrin modulation, followed by the recruitment of endothelial cells (VEGFR2-positive) to the distant site [36].
Cancer cells and the tumor microenvironment produce factors that influence angiogenic processes, with the key drivers being VEGF-A [37,38] binding to VEGFR2 receptor [39]. Alterations of protooncogenes (RAS and SRC) and tumor suppressor genes (TP53 and VHL) correlate with VEGF overproduction by tumor cells. Hypoxia is the principal stimulator of VEGF production, and hypoxia-inducible transcription factors (HIF-1α and HIF-2α) play a central role in VEGF regulation. Other angiogenesis inductors, such as FGF1, EGF2, PDGF-B, PDGF-C, and EGF, bind to their respective receptors on blood vessel endothelial cells and induce proliferation and migration [40]. Besides the conventional angiogenic mediators, BMP9 signaling and Shh signaling also participate in the process [41]. In addition, exosomes released by cancer and immune cells may transport various proangiogenic molecules like VEGF, MMPs, and microRNAs [42].

3. The Relationship between Microbiome and Cancer Progression-Related Processes

In recent years, the correlation between the microbiome, cancer, and metastatic disease has gained more attention (Figure 2). Many studies confirmed that certain microbes and their metabolites are associated with a better/worse therapy response and patient outcomes.
Understanding the mechanisms by which unfavorable microbes have an impact on tumor progression is an active area of recent research. Therefore, intensive research in numerous ongoing clinical trials might shed more light on prognostic microbial markers for treatment outcomes in metastatic disease (Table 1). The identification of microbial biomarkers will help to understand how the microbiome is implicated in cancer progression.

3.1. Microbiome in Epithelial-to-Mesenchymal Transition

The association between specific microbiome compositions, the inflammatory response, and therapy resistance related to EMT is still under debate [43]. Alterations in the gut microbiome might support EMT via the TGFβ, WNT, and Notch signaling pathways, and Slug, SNAIL, Twist, ZEB1, and ZEB2 transcription factors resulted in invasive and metastatic cancer processes [44,45,46]. A pathological microbiome in pancreatic ductal adenocarcinoma (PDAC) patients promoted EMT via the activation of transcription factors, including TGFβ and TNFα [47]. Antibiotic-induced gut dysbiosis supported macrophage activation and the production of inflammatory cytokines, subsequently promoting EMT in colorectal cancer (CRC) [48]. Fusobacterium nucleatum, Enterococcus faecalis, Bacteroides fragilis, Escherichia coli, and Salmonella enterica are known microbes involved in CRC progression, producing virulence factors that contribute to EMT and cancer progression [49]. F. nucleatum-infected CRC cells supported the EMT cell phenotype and elevated cancer cell migration, as well as tumorsphere formation [50]. Similarly, EMT-related morphological changes and the upregulation of mesenchymal marker ZEB1 were documented in cag pathogenicity island+ H. pylori-infected gastric epithelial cells [51]. Marques et al. noted that the H. pylori infection of gastric cells reduced Afadin and increased ZEB1, vimentin, Slug, Snail, and N-cadherin. Since Afadin regulates adherens and tight junctions, its loss due to bacterial infection might play a role in aggressive gastric cancer phenotypes [52].
The tumor-associated butyrate-producing bacterium SM4/1, E. coli str. K-12 substrate MG1655, Saccharomonospora viridis DSM 43,017, and E. coli O157:H7 str. EC4115 correlated with the expression of EMT-related genes (TGFB, RhoA, vimentin-associated genes, SNAI2, SNAI3, and TWIST1) in muscle-invasive bladder cancer. In addition, the level of both E. coli O157:H7 str. EC4115 and Oscillatoria sp. CCAP 1459/13 was negatively associated with E-cadherin expression [53]. In the T24 cell line, infection with E. coli significantly upregulated vimentin, reactive oxygen species levels, and stemness markers (CD44, NANOG, SOX2, and OCT4), while it downregulated CK19. The authors observed that E. coli-infected bladder cancer cells had a specific elongated morphology with a shortage of contact between cells, while non-infected cells had an adhesive ability with epithelial morphology [54]. Gingival squamous cell carcinoma tissue samples showed the presence of Porphyromonas gingivalis [55]. Porphyromonas supports the invasion and metastasis of oral cancer cells via EMT processes. The exposure of cancer cells to P. gingivalis for a long period promoted migration, invasiveness, and resistance to chemotherapeutics [56,57]. The infection of gingival epithelial cells with P. gingivalis increased ZEB1 expression, leading to EMT regulation. Conversely, bacterial strains lacking FimA did not promote the expression of ZEB1 [58].
Serum levels of bacterial lipopolysaccharide (LPS) were found to be elevated in individuals diagnosed with esophageal cancer. An in vitro study by Peng et al. revealed an increase in N-cadherin and vimentin in EC109 cells after treatment with LPS, accompanied by reduced E-cadherin levels. The achieved results suggest the potential role of LPS in promoting migration, invasion, and EMT initiation [59]. Microbiota might control cancer stem cells via miRNA and circRNA regulation [60]. Broad-spectrum antibiotic treatment upregulated the level of mmu_circ_0000730, showing its association with cancer cell stemness and EMT via an upregulated SOX9 expression level. As reported, siRNA targeted mmu_circ_0000730-inhibited invasive/migrative processes and reduced metastasis development [61]. Yan et al. confirmed the involvement of F. nucleatum in cancer stem cells and EMT crosstalk in cancer progression. The expression levels of EMT markers (E-cadherin and N-cadherin) and a cancer stem cell marker (Nanog) correlated with the abundance of F. nucleatum in CRC tissue samples [62]. Previous data observed that infection with Fusobacterium upregulated miR-21 levels, and the upregulation of miR-21 correlated with distant metastases and cancer stage [63]. In breast cancer, bacterial metabolites, such as short-chain fatty acids (SCFAs), lithocholic acid, cadaverine, and indole derivatives, exert an influence on the EMT process [64,65]. Ujlaki et al. screened the antineoplastic properties of bacterial-associated metabolites. The application of 3-hydroxyphenyl acetic acid, 4-hydroxybenzoic acid, and vanillic acid led to a hyperproliferative impact on breast cancer cells, while the application of butyric acid, glycolic acid, d-mannitol, 2,3-butanediol, and trans-ferulic acid had cytostatic effects. Anti-EMT properties were documented in the case of applied 3-hydroxyphenyl acetic acid, 4-hydroxybenzoic acid, 2,3-butanediol, and hydrocinnamic acid [65]. The treatment of breast cancer cells with indolepropionic acid, a microbiota-derived tryptophan metabolite, reverted EMT [66].

3.2. Microbiome and Angiogenesis

Tumor angiogenesis is involved not only in tumor growth but also in tumor progression and metastasis. According to findings, microbiota plays a role in angiogenesis via VEGF and inflammatory cells [67]. Studies showed that microbiota-derived SCFAs may have an impact on angiogenesis. Butyrate, a major member of SCFAs, plays a role in many cellular processes. Decreased levels of sodium butyrate supported angiogenesis [68]. Lithocholic acid is a bacterial metabolite, and it plays a role as a tumor promoter in human CRC cells [69]. Enterotoxin produced by enterotoxigenic E. coli might activate a cGMP-dependent signaling pathway that decreases VEGF and VCAM-1, both associated with angiogenesis and tumor metastasis [70]. Trimethylamine N-oxide (TMAO), as a microbiome-derived metabolite, increased CRC cell proliferation in vitro. Moreover, in vivo experiments confirmed higher levels of TMAO in the circulation, an increased tumor volume, and promoted angiogenesis in choline-fed CRC mice [71]. Changes in the gut microbiome participate in the transformation of adenomas into carcinomas. These alterations resulted from reduced butyrate levels and the secretion of mutagenic metabolites, including spermidine and trimethylamine. Therefore, the CRC microbiome might promote inflammation, angiogenesis, and apoptosis via increased/decreased histamine, glutamine, and pyruvate levels [72]. The crosstalk between the microbiome, macrophages, and cancer cells is still unrevealed. CRC-derived exosomes can interplay with macrophages within the tumor microenvironment and participate in angiogenesis. Moreover, macrophages activated by microbiome-derived exosomes might induce inflammation [73]. Feces transferred from CRC patients into a model of germ-free and azoxymethane-treated mice upregulated genes associated with angiogenesis, invasion, metastasis, and proliferation [74]. Four quorum-sensing peptides from Bacillus subtilis (PhrG), Streptococcus mitis (CSP), and E. coli (EDF) with its tripeptide analog NWN promoted angiogenesis and breast cancer cell invasion [75]. Similarly, the bacteria-produced quorum-sensing peptides such as EntF-metabolite from Enterococcus faecium, Phr0662 from Bacillus spp., and EDF derived from E. coli stimulated colon cancer cell invasion and angiogenesis [76].

4. The Studies of Microbiome Composition in Metastatic Disease

Recently, Hilmi et al. studied samples obtained from the lymph nodes, lungs, and livers of patients suffering from different cancer types, such as breast, lung, and colorectal malignancies. A higher presence of F. nucleatum was specific to lung metastases. The microbial load in lymph node metastases was lower than in liver and lung metastases. However, the authors did not observe a relationship between the type of primary tumor and the microbial composition in metastases [77]. The level of Eubacterium halli in stool samples is negatively associated with fatigue in patients with advanced, metastatic, unresectable colon, ovarian, cervical, and non-small-cell lung cancers [78]. In vivo experiments confirmed that gut microbial depletion via a broad-spectrum antibiotic cocktail reduced the incidence of metastases in melanoma, pancreatic, or colon cancer murine models [79]. Spakowicz et al. performed a retrospective analysis of 690 patients treated with immunotherapy for metastatic melanoma or non-small-cell lung cancer. The results showed that antibiotics and corticosteroids reduced overall survival (OS), but no direct microbiome measurements were performed [80].

4.1. Gastrointestinal Malignancies

Gut dysbiosis and intestinal inflammation represent critical factors in CRC progression [81]. Many studies documented differences in the gut microbiome composition between early and definitive CRC stages [82,83]. Higher levels of F. nucleatum in fecal and tumor samples correlated with advanced-stage CRC in patients. This microorganism activated tumor-derived CCL20 expression, increased macrophage infiltration, induced M2 polarization, and enhanced metastasis [84]. Similarly, F. nucleatum was more abundant in feces from patients with positive lymph node metastases. In vitro experiments showed that bacterial infection increased the migration of HCT-116 and LoVo cells. However, the heat-killed bacterium could not support cell migration. Accordingly, an in vivo analysis showed that an injection of Fusobacterium-treated cells induced lung metastatic nodules in nude mice [85]. Fusobacterium might contribute to CRC metastases via declined m6A modifications in CRC cells and patient-derived xenografts, which resulted in more aggressive cancer cell phenotypes [86]. The presence of Fusobacterium in primary colon adenocarcinoma correlated with Bacteroides, Selenomonas, and Prevotella in distant metastases. Viable Fusobacterium was detected in murine xenografts, and metronidazole administration significantly reduced intratumoral Fusobacterium load and tumor growth [87].
Wang et al. assessed the effect of combined regorafenib plus toripalimab in patients with refractory metastatic CRC. Patients not responding to combined treatment were reported to have an increased amount of F. nucleatum. A lower response rate was documented in patients with liver metastases [88]. However, supplementation with Faecalibacterium prausnitznii could reverse CRC progression via several mechanisms and reduce inflammation [89]. In mice, a colonic bacterial biofilm composed of E. coli and B. fragilis was responsible for faster CRC onset, disease progression, and mortality than colonization only with one bacterial strain alone [90]. The B. fragilis toxin is responsible for lost cell adhesion resulting in EMT [91]. Changes in the EMT-associated genes SNAI1 and VIM were documented after the co-cultivation of Caco-2 cells with bacteria from CRC biopsies. The downregulation of SNAI1 was observed in all affected cells. However, VIM expression changes depended on co-cultivation with specific bacteria. Proteus vulgaris increased VIM expression, but Proteus mirabilis and Pseudomonas aeruginosa were associated with reduced expression [92]. Sun et al. assessed the effect of the Quxie capsule (QX) as herbal medicine on the gut microbiome in patients with unresectable metastatic CRC. A decreased amount of Actinobacteria became elevated after QX treatment. An LEfSe analysis showed that the microbiome before QX treatment was composed of c_Gammaproteobacteria and g_Lachnospiraceae UCG 008, while f_Ruminococcaceae and g_Peptostreptococcus dominated after QX treatment. Moreover, high levels of CD4 and CD8 cells were observed in the QX-treated group [93].
The poor prognosis of pancreatic cancer reflects the necessity of novel non-invasive markers in the management of patients suffering from metastatic or locally advanced pancreatic cancer. Therefore, a fecal microbiome analysis might help to determine potential microbiome-related biomarkers in the early diagnosis of patients [94]. Jeong et al. investigated the microbiome in tumor and normal adjacent tissue samples from patients with PDAC. A higher number of metastases in lymph nodes correlated with a higher abundance of Comamonas and Turicibacter in tumor tissues, while there was a lower amount of Tepidimonas, Enhydrobacter, Turicibacter, and Wautersiella in normal tissues. Alpha diversity was higher in patients with cancer recurrence with low levels of Streptococcus and Akkermansia in tumor samples [95].
The administration of a nanoparticle conjugation of ginsenoside Rg3 (NpRg3) reduced Firmicutes and increased Bacteroidetes and Verrucomicrobia in the dimethylnitrosamine-induced hepatocellular carcinoma murine model. The results confirmed that NpRg3 administration suppressed the development of lung metastases. Although circulating tumor cells from primary tumors are the main factor affecting cancer metastasis, NpRg3 might inhibit metastatic spreading via the reduced mobilization of endothelial progenitor cells in the circulation [96].

4.2. Lung Cancer

Huang et al. observed that bronchial washing fluid might characterize the microbial composition of lung tumor tissues. Higher amounts of Veillonella, Megasphaera, Actinomyces, and Arthrobacter genera were documented in a group of lung adenocarcinoma patients without distant metastases than in squamous cell lung carcinoma patients without metastases. However, lower levels of Capnocytophaga and Rothia were found in lung adenocarcinoma with distant metastases than in squamous cell lung carcinoma with distant metastases. Specifically, the Streptococcus genus was more reduced in lung adenocarcinoma with metastases than in lung adenocarcinoma without metastases. The results confirmed that the microbial composition differed in histologic types of lung cancer with or without metastases [97]. Another microbiome study showed higher Legionella in lung tissues from patients who developed metastases, while Thermus was more abundant in tissue samples from patients with advanced-stage (IIIB and IV) lung cancer [98]. A respiratory tract infected with Streptococcus pneumoniae promoted the development of liver metastases in the murine model via TLR2 activation [99]. Higher exposure to cigarette smoke and non-typeable Haemophilus influenzae supported lung metastasis growth in the lung cancer model. The authors noted that cigarette smoke caused the disruption of pulmonary barrier integrity, allowing for the translocation of bacterial factors into tumors [100].

4.3. Breast Cancer

Some studies highlighted the differences in the microbiome composition between samples from breast tumors and non-malignant breast tissues [101]. For the first time, Buchta Rosean and a research team documented that dysbiotic changes in the gut microbiome increased the dissemination process in the HR+ breast cancer murine model. Antibiotic pretreatment caused changes at the phylum and genus levels. The amounts of Alistipes, Blautia, Parabacteroides, and Escherichia/Shigella genera were elevated in mice with antibiotic-induced commensal dysbiosis. Antibiotic-induced dysbiosis enhanced tumor cell dissemination in peripheral blood, lungs, and axillary lymph nodes [102]. An analysis of fecal samples revealed that Acinetobacter, Bacilli, Campylobacter, Collinsella, Lactobacillales, Veillonella, Streptococcus, Epsilonproteobacteria, Pseudomonadales, and Moraxellaceae were more abundant in the gut microbiome of breast cancer patients with bone metastases than in control samples. However, the analysis revealed significantly decreased amounts of Megamonas, Clostridia, Akkermansia, Gemmiger, and Paraprevotella in the fecal samples of metastatic patients. Therefore, the authors suggested that lower levels of Megamonas and Akkermansia might correlate with the presence of bone metastases [103]. The tumor microbiome depletion induced by antibiotic treatment inhibited lung metastasis development in the murine spontaneous breast-tumor model. As detected, the intratumoral administration of specific bacteria, such as Staphylococcus xylosus, Ligilactobacillus animalis, and Spilopsyllus cuniculi, elevated the number of lung metastases without affecting primary tumors in mice [11].
Parhi et al. confirmed the pro-metastatic effect of F. nucleatum in the murine model. A tail vein microbe injection supported the progression of AT3 mammary tumors and the development of lung metastases. Mammary tumors were colonized by Fusobacterium via bacterial attachment to display Gal-GalNAc on tumor cells [104]. Zhu et al. observed that the administration of broad-spectrum antibiotics promoted cancer metastases and decreased the survival rate, showing low Bifidobacterium in antibiotic-treated mice. However, fecal microbiota transplantation (FMT) from SPF mice caused the restoration of the gut microbiome composition [61]. Neoadjuvant chemotherapy decreased the intratumoral level of Streptococcus while elevating the amount of Pseudomonas. A higher abundance of Brevundimonas and Staphylococcus was reported in primary breast tumors from women with distant metastases. In vitro experiments showed that breast cancer cells treated with 10% P. aeruginosa-conditioned media decreased doxorubicin-mediated cleaved caspase 7 in breast cancer cells [105].

4.4. Head and Neck Cancers

Eun et al. documented statistically significant differences in the oral microbiome between patients with and without metastases in oral squamous cell carcinoma. An analysis of the oral microbiome in saliva samples from patients with tumors in the oral cavity showed that Tannerella and Fusobacterium genera were more abundant in participants without metastases, while Prevotella, Stomatobaculum, Bifidobacterium, Peptostreptococcaceae, Shuttleworthia, and Finegoldia had a higher presence in metastatic patients [106].
A study with 802 nasopharyngeal cancer patients revealed that a higher bacterial load within tumors correlated with worse metastasis-free survival and patient OS. The predominant bacterial taxa in the tumor samples were Corynebacterium and Staphylococcus [107]. Yuan et al. revealed specific intratumoral bacterial taxa, including Pseudomonas, Rhodococcus, Sphingomonas, Streptococcus, Granulicatella, Haemophilus, and Coriobacteriales, which predicted papillary thyroid cancer invasiveness. A higher alpha diversity was documented in tumor samples from patients with advanced stages than in patients with lower disease stages. The interaction between the tumor microbiome and thyroid hormones might contribute to cancer progressiveness, but more research is needed in this area [108].

4.5. Genitourinary Cancers

A study of the bacterial composition of fecal samples from patients with advanced metastatic castrate-resistant prostate cancer treated with immunotherapy showed differences between responders and non-responders to pembrolizumab. Bacteroides thetaiotaomicron, B. fragilis, and Roseburia unassigned dominated in responder samples. In contrast to other studies showing that the presence of favorable Akkermansia muciniphila correlated with a better anti-cancer treatment response, Peiffer et al. observed its lower amount in responder fecal samples [109]. The level of intratumoral microbes, including Methylobacterium radiotolerans JCM 2831, Delftia acidovorans SPH-1, Rhodococcus erythropolis PR4, Stenotrophomonas maltophilia K279a, and Meiothermus silvanus DSM 9946, negatively correlated with the tumor-node-metastasis stage in patients with prostate cancer [110].
A higher fecal microbial diversity and A. muciniphila enrichment were associated with a better clinical response to nivolumab or nivolumab plus ipilimumab in patients with metastatic renal cell carcinoma [111]. However, an analysis of fecal samples from patients with metastatic renal cell carcinoma noted that Akkermansia was not responsible for treatment efficacy due to its detection in the gut microbiome of seven responders and six non-responders. Differences in the baseline gut microbiome between responding and non-responding patients were not supposed to predict patient outcomes [112]. Probiotic supplementation elevated the level of Bifidobacterium spp. in patients with metastatic renal cell carcinoma. A clinical benefit from targeted therapy using vascular endothelial growth factor-tyrosine kinase inhibitors was observed in patients enriched with Barnesiella intestinihominis and A. muciniphila in the microbiome [113]. Antibiotic supplementation targeting Bacteroides spp. in the gut microbiome improved progression-free survival (PFS) in patients with metastatic renal cell carcinoma who received VEGF-tyrosine kinase inhibitors (TKIs) [114].

4.6. Metastatic Melanoma

Oral and gut microbial communities were diverse in 112 patients with metastatic melanoma. Significant enrichments of Bacteroidales in the fecal microbiome and Lactobacillales in the oral microbiome were documented in patient samples using 16S rRNA sequencing. In the case of response to programed cell death protein 1 (PD-1) blockade, alpha diversity was higher in the responders’ fecal microbiome, but differences in the oral microbiome were not observed between responders and non-responders to therapy [115]. Mizuhashi et al. found an association between the level of the genus Corynebacterium in swab samples and melanoma progression. The results showed that Corynebacterium was present in 76.9% and 28.6% of patients at stages III/IV and I/II, respectively [116]. A microbiome analysis of fecal samples from 42 metastatic melanoma patients before immunotherapy uncovered an enrichment of Bifidobacterium longum, Collinsella aerofaciens, and E. faecium in the gut microbiome of responding patients, whereas Ruminococcus obeum and Roseburia intestinalis were more abundant in non-responders [117]. The gut microbiome of metastatic melanoma patients responding to PD-1 blockade was enriched in Clostridiales. On the contrary, a higher amount of Bacteroidales dominated in non-responding patients [118]. Vitali et al. found that Prevotella, Clostridium IV, Holdemania, and Anaerofustis were abundant in melanoma patients’ gut microbiome. According to the authors, a richer gut microbiome was shown in patients with stage I/II melanoma than in patients with metastatic disease [119].
The fundamental studies focusing on the microbiome composition in metastatic disease are summarized in Table 2.

5. Microbiota Modulation and Cancer Progression

Gut microbiome modulation leading to increased intestinal barrier and anti-inflammatory responses might inhibit pro-tumorigenic processes, including cancer progression, migration, invasion, and angiogenesis [120,121]. The intragastric administration of Lactobacillus reuteri FLRE5K1 inhibited the incidence of tumors in BALB/C mice injected with melanoma cells. The results indicated that L. reuteri FLRE5K1 might restrain the development of tumors due to the blockade of the migration and colonization of cancer cells [122]. A preclinical study noted that fecal transplants from obese mice to lean mice with B16F10 tumors stimulated melanoma development and supported cancer progression [123]. The aim of recent studies is to assess the effect of probiotic supplementation on tumor progression.
Chen et al. showed that probiotics composed of B. longum, Bifidobacterium breve, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus casei, Lactobacillus bulgaricus, and Streptococcus thermophilus attenuated the development of lung metastases and prolonged survival in melanoma-bearing mice. Probiotic supplementation led to increased levels of Lachnospiraceae, Streptococcus, and Lachnoclostridium [124]. Aerosolized probiotic Lactobacillus rhamnosus, which reached lung murine tissue, reduced the number of lung metastases. Moreover, aerosolized L. rhamnosus and Bifidobacterium bifidum increased the effect of conventional chemotherapeutic dacarbazine in melanoma-bearing mice [125]. A Prohep probiotic product composed of L. rhamnosus GG, E. coli Nissle 1917, and VSL#3 (1:1:1) orally administered to mice bearing hepatocellular carcinoma retarded tumor growth and inhibited angiogenesis. Prohep administration altered the gut microbiome toward valerate producer Oscillibacter and propionate producer Prevotella, both known for their ability to reduce Th17 polarization and support Treg/Tr1 cells in the gut. The results showed that probiotics reduced the recruitment of Th17 cells, which secrete pro-inflammatory cytokines to the tumor microenvironment. Additionally, several angiogenic factors, including ANG2, FLT-1, KDR, TEK, and VEGF-A, were downregulated in the probiotic-supplemented group [126]. Daily oral administration of CBM588 containing Clostridium butyricum prolonged PFS in patients with metastatic renal cell carcinoma treated with nivolumab and ipilimumab. The level of Bifidobacterium spp. was higher in probiotic-supplemented patients who responded to immunotherapy, while the results showed a decline in Desulfovibrio spp. However, the toxicity rate was not different between the supplemented and control groups [127]. A probiotic mixture of eight bacterial strains mitigated the length and severity of chemotherapy-associated diarrhea in CRC animal models with liver metastases. Moreover, probiotics support gastrointestinal regeneration after chemotherapeutic treatment [128]. Shang et al. documented that an intragastric probiotic mixture of B. longum, B. bifidum, L. acidophilus, and L. plantarum attenuated cancer cell proliferation and even the development of metastasis in mouse models of CRC [129]. Baruch et al. performed FMT from 2 selected donors previously treated with immunotherapy for metastatic melanoma into 10 recipients with confirmed progression on PD–1 blockade. The presence of favorable Lachnospiraceae was observed in both donors. The feces transfer from donors caused a shift in the gut microbiome in metastatic melanoma recipients with abundant favorable Veillonellaceae and a decline in B. bifidum [130].

6. Conclusions and Future Directions

The gut and intratumoral microbiomes can influence cancer progression and metastatic processes in various ways, including inducing inflammation and immune system modulation, affecting metabolism and providing energy for cancer cell spread, promoting the angiogenesis caused by microbial metabolites, and impacting cancer treatment efficacy to control metastatic disease. Particular attention should be paid to addressing the ability of specific microorganisms and microbiota-derived metabolites to shape the immune system and tumor microenvironment, potentially promoting the growth and spread of cancer cells. Longitudinal multi-center clinical trials targeting the microbiome composition and function in cancer patients before, during, and after treatment would help uncover distinctive microbiome-associated patterns and their effects on recurrence and survival rates. However, the standardization of methods and procedures for microbiome analyses will be crucial to obtain the most relevant results for clinical use.
Mounting evidence reported a significant correlation between the efficacy of immunotherapy and the gut microbiome composition in patients with metastatic disease. Tumor microbiome profiling along with gut microbiome characterization in individual patients could lead to future personalized therapy to increase the efficacy and decrease the toxicity of anti-cancer treatment. Furthermore, the tumor and gut microbiomes could undergo similar evolution, and the microbiome composition could be assessed at several time points. Another important field of research includes therapeutic interventions aimed at modifying the tumor and/or gut microbiome, including different approaches, such as antibiotic therapy; the modification of the tumor microenvironment; treatments affecting immune cells; and the colonization of beneficial bacteria by prebiotics, probiotics, and fecal microbiome transplant. Further research is required to obtain more evidence elucidating the mechanisms and signaling pathways behind the microbiota–host interactions that drive metastasis and malignant progression. A deeper understanding could help integrate microbiome-based interventions into clinical practice for improving cancer patient outcomes.

Author Contributions

Conceptualization, S.C.; writing—original draft preparation, A.S., S.C. and B.M.; writing—review and editing, M.M., B.M. and S.C.; visualization, A.S. and S.C.; project administration, S.C. and B.M.; funding acquisition, S.C. and B.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Scientific Grant Agency of the Ministry of Education, Science, Research and Sport of the Slovak Republic and Slovak Academy of Sciences (VEGA) contract Nos. 2/0069/22 and 1/0738/21. The funding source had no influence on the writing of the manuscript.

Acknowledgments

Figure 1 and Figure 2, and the Graphical Abstract were created with Biorender.com. Information for this Review was compiled by searching the PubMed and MEDLINE databases. Only articles published in English were considered. The search terms included “gut and tumor microbiome” and “gut microbiota” in association with the search terms “cancer progression” or “metastasis” or “angiogenesis” or “epithelial-to-mesenchymal transition”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sevcikova, A.; Izoldova, N.; Stevurkova, V.; Kasperova, B.; Chovanec, M.; Ciernikova, S.; Mego, M. The Impact of the Microbiome on Resistance to Cancer Treatment with Chemotherapeutic Agents and Immunotherapy. Int. J. Mol. Sci. 2022, 23, 488. [Google Scholar] [CrossRef] [PubMed]
  2. Cullin, N.; Azevedo Antunes, C.; Straussman, R.; Stein-Thoeringer, C.K.; Elinav, E. Microbiome and cancer. Cancer Cell 2021, 39, 1317–1341. [Google Scholar] [CrossRef] [PubMed]
  3. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  4. Massague, J.; Ganesh, K. Metastasis-Initiating Cells and Ecosystems. Cancer Discov. 2021, 11, 971–994. [Google Scholar] [CrossRef] [PubMed]
  5. Gerstberger, S.; Jiang, Q.; Ganesh, K. Metastasis. Cell 2023, 186, 1564–1579. [Google Scholar] [CrossRef] [PubMed]
  6. Gao, F.; Yu, B.; Rao, B.; Sun, Y.; Yu, J.; Wang, D.; Cui, G.; Ren, Z. The effect of the intratumoral microbiome on tumor occurrence, progression, prognosis and treatment. Front. Immunol. 2022, 13, 1051987. [Google Scholar] [CrossRef] [PubMed]
  7. Galeano Nino, J.L.; Wu, H.; LaCourse, K.D.; Kempchinsky, A.G.; Baryiames, A.; Barber, B.; Futran, N.; Houlton, J.; Sather, C.; Sicinska, E.; et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature 2022, 611, 810–817. [Google Scholar] [CrossRef]
  8. Liu, J.; Luo, F.; Wen, L.; Zhao, Z.; Sun, H. Current Understanding of Microbiomes in Cancer Metastasis. Cancers 2023, 15, 1893. [Google Scholar] [CrossRef]
  9. Hofman, P.; Vouret-Craviari, V. Microbes-induced EMT at the crossroad of inflammation and cancer. Gut Microbes 2012, 3, 176–185. [Google Scholar] [CrossRef]
  10. Caven, L.T.; Brinkworth, A.J.; Carabeo, R.A. Chlamydia trachomatis induces the transcriptional activity of host YAP in a Hippo-independent fashion. Front. Cell Infect. Microbiol. 2023, 13, 1098420. [Google Scholar] [CrossRef]
  11. Fu, A.; Yao, B.; Dong, T.; Chen, Y.; Yao, J.; Liu, Y.; Li, H.; Bai, H.; Liu, X.; Zhang, Y.; et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell 2022, 185, 1356–1372.e1326. [Google Scholar] [CrossRef] [PubMed]
  12. Brandt, S.; Kwok, T.; Hartig, R.; Konig, W.; Backert, S. NF-κB activation and potentiation of proinflammatory responses by the Helicobacter pylori CagA protein. Proc. Natl. Acad. Sci. USA 2005, 102, 9300–9305. [Google Scholar] [CrossRef] [PubMed]
  13. Yin, Y.; Grabowska, A.M.; Clarke, P.A.; Whelband, E.; Robinson, K.; Argent, R.H.; Tobias, A.; Kumari, R.; Atherton, J.C.; Watson, S.A. Helicobacter pylori potentiates epithelial:mesenchymal transition in gastric cancer: Links to soluble HB-EGF, gastrin and matrix metalloproteinase-7. Gut 2010, 59, 1037–1045. [Google Scholar] [CrossRef] [PubMed]
  14. Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct. Target. Ther. 2020, 5, 28. [Google Scholar] [CrossRef] [PubMed]
  15. Thiery, J.P. Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2002, 2, 442–454. [Google Scholar] [CrossRef] [PubMed]
  16. Yang, J.; Antin, P.; Berx, G.; Blanpain, C.; Brabletz, T.; Bronner, M.; Campbell, K.; Cano, A.; Casanova, J.; Christofori, G.; et al. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2020, 21, 341–352. [Google Scholar] [CrossRef] [PubMed]
  17. Giuliano, M.; Giordano, A.; Jackson, S.; Hess, K.R.; De Giorgi, U.; Mego, M.; Handy, B.C.; Ueno, N.T.; Alvarez, R.H.; De Laurentiis, M.; et al. Circulating tumor cells as prognostic and predictive markers in metastatic breast cancer patients receiving first-line systemic treatment. Breast Cancer Res. 2011, 13, R67. [Google Scholar] [CrossRef]
  18. Mego, M.; Karaba, M.; Minarik, G.; Benca, J.; Silvia, J.; Sedlackova, T.; Manasova, D.; Kalavska, K.; Pindak, D.; Cristofanilli, M.; et al. Circulating Tumor Cells with Epithelial-to-mesenchymal Transition Phenotypes Associated with Inferior Outcomes in Primary Breast Cancer. Anticancer Res. 2019, 39, 1829–1837. [Google Scholar] [CrossRef]
  19. Fridrichova, I.; Kalinkova, L.; Ciernikova, S. Clinical Relevancy of Circulating Tumor Cells in Breast Cancer: Epithelial or Mesenchymal Characteristics, Single Cells or Clusters? Int. J. Mol. Sci. 2022, 23, 12141. [Google Scholar] [CrossRef]
  20. Kim, M.Y.; Oskarsson, T.; Acharyya, S.; Nguyen, D.X.; Zhang, X.H.; Norton, L.; Massague, J. Tumor self-seeding by circulating cancer cells. Cell 2009, 139, 1315–1326. [Google Scholar] [CrossRef]
  21. Comen, E.; Norton, L. Self-seeding in cancer. Recent Results Cancer Res. 2012, 195, 13–23. [Google Scholar] [CrossRef]
  22. Park, J.; Wysocki, R.W.; Amoozgar, Z.; Maiorino, L.; Fein, M.R.; Jorns, J.; Schott, A.F.; Kinugasa-Katayama, Y.; Lee, Y.; Won, N.H.; et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl. Med. 2016, 8, 361ra138. [Google Scholar] [CrossRef] [PubMed]
  23. Cools-Lartigue, J.; Spicer, J.; McDonald, B.; Gowing, S.; Chow, S.; Giannias, B.; Bourdeau, F.; Kubes, P.; Ferri, L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J. Clin. Investig. 2013, 123, 3446–3458. [Google Scholar] [CrossRef] [PubMed]
  24. Zhong, W.; Wang, Q.; Shen, X.; Du, J. The emerging role of neutrophil extracellular traps in cancer: From lab to ward. Front. Oncol. 2023, 13, 1163802. [Google Scholar] [CrossRef] [PubMed]
  25. Adrover, J.M.; McDowell, S.A.C.; He, X.Y.; Quail, D.F.; Egeblad, M. NETworking with cancer: The bidirectional interplay between cancer and neutrophil extracellular traps. Cancer Cell 2023, 41, 505–526. [Google Scholar] [CrossRef] [PubMed]
  26. Hu, W.; Lee, S.M.L.; Bazhin, A.V.; Guba, M.; Werner, J.; Niess, H. Neutrophil extracellular traps facilitate cancer metastasis: Cellular mechanisms and therapeutic strategies. J. Cancer Res. Clin. Oncol. 2023, 149, 2191–2210. [Google Scholar] [CrossRef]
  27. Nepali, P.R.; Kyprianou, N. Anoikis in phenotypic reprogramming of the prostate tumor microenvironment. Front. Endocrinol. 2023, 14, 1160267. [Google Scholar] [CrossRef] [PubMed]
  28. Paoli, P.; Giannoni, E.; Chiarugi, P. Anoikis molecular pathways and its role in cancer progression. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2013, 1833, 3481–3498. [Google Scholar] [CrossRef]
  29. Pretzsch, E.; Bosch, F.; Neumann, J.; Ganschow, P.; Bazhin, A.; Guba, M.; Werner, J.; Angele, M. Mechanisms of Metastasis in Colorectal Cancer and Metastatic Organotropism: Hematogenous versus Peritoneal Spread. J. Oncol. 2019, 2019, 7407190. [Google Scholar] [CrossRef]
  30. Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
  31. Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef] [PubMed]
  32. Huang, H. Matrix Metalloproteinase-9 (MMP-9) as a Cancer Biomarker and MMP-9 Biosensors: Recent Advances. Sensors 2018, 18, 3249. [Google Scholar] [CrossRef] [PubMed]
  33. Kryczek, I.; Wei, S.; Keller, E.; Liu, R.; Zou, W. Stroma-derived factor (SDF-1/CXCL12) and human tumor pathogenesis. Am. J. Physiol. Cell Physiol. 2007, 292, C987–C995. [Google Scholar] [CrossRef] [PubMed]
  34. Xu, C.; Zhao, H.; Chen, H.; Yao, Q. CXCR4 in breast cancer: Oncogenic role and therapeutic targeting. Drug Des. Dev. Ther. 2015, 9, 4953–4964. [Google Scholar] [CrossRef]
  35. Muller, A.; Homey, B.; Soto, H.; Ge, N.; Catron, D.; Buchanan, M.E.; McClanahan, T.; Murphy, E.; Yuan, W.; Wagner, S.N.; et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 2001, 410, 50–56. [Google Scholar] [CrossRef] [PubMed]
  36. Kaplan, R.N.; Rafii, S.; Lyden, D. Preparing the “soil”: The premetastatic niche. Cancer Res. 2006, 66, 11089–11093. [Google Scholar] [CrossRef] [PubMed]
  37. De Palma, M.; Biziato, D.; Petrova, T.V. Microenvironmental regulation of tumour angiogenesis. Nat. Rev. Cancer 2017, 17, 457–474. [Google Scholar] [CrossRef] [PubMed]
  38. Kim, H.J.; Ji, Y.R.; Lee, Y.M. Crosstalk between angiogenesis and immune regulation in the tumor microenvironment. Arch. Pharm. Res. 2022, 45, 401–416. [Google Scholar] [CrossRef]
  39. Zirlik, K.; Duyster, J. Anti-Angiogenics: Current Situation and Future Perspectives. Oncol. Res. Treat. 2018, 41, 166–171. [Google Scholar] [CrossRef]
  40. Ornitz, D.M.; Itoh, N. The Fibroblast Growth Factor signaling pathway. Wiley Interdiscip. Rev. Dev. Biol. 2015, 4, 215–266. [Google Scholar] [CrossRef]
  41. Harry, J.A.; Ormiston, M.L. Novel Pathways for Targeting Tumor Angiogenesis in Metastatic Breast Cancer. Front. Oncol. 2021, 11, 772305. [Google Scholar] [CrossRef] [PubMed]
  42. Olejarz, W.; Kubiak-Tomaszewska, G.; Chrzanowska, A.; Lorenc, T. Exosomes in Angiogenesis and Anti-Angiogenic Therapy in Cancers. Int. J. Mol. Sci. 2020, 21, 5840. [Google Scholar] [CrossRef] [PubMed]
  43. Roche, J. The Epithelial-to-Mesenchymal Transition in Cancer. Cancers 2018, 10, 52. [Google Scholar] [CrossRef] [PubMed]
  44. Umar, S. Enteric pathogens and cellular transformation: Bridging the gaps. Oncotarget 2014, 5, 6573–6575. [Google Scholar] [CrossRef] [PubMed]
  45. Thiery, J.P.; Acloque, H.; Huang, R.Y.; Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef] [PubMed]
  46. Chandrakesan, P.; Roy, B.; Jakkula, L.U.; Ahmed, I.; Ramamoorthy, P.; Tawfik, O.; Papineni, R.; Houchen, C.; Anant, S.; Umar, S. Utility of a bacterial infection model to study epithelial-mesenchymal transition, mesenchymal-epithelial transition or tumorigenesis. Oncogene 2014, 33, 2639–2654. [Google Scholar] [CrossRef] [PubMed]
  47. Basu, M.; Philipp, L.M.; Baines, J.F.; Sebens, S. The Microbiome Tumor Axis: How the Microbiome Could Contribute to Clonal Heterogeneity and Disease Outcome in Pancreatic Cancer. Front. Oncol. 2021, 11, 740606. [Google Scholar] [CrossRef]
  48. Wan, G.; Xie, M.; Yu, H.; Chen, H. Intestinal dysbacteriosis activates tumor-associated macrophages to promote epithelial-mesenchymal transition of colorectal cancer. Innate Immun. 2018, 24, 480–489. [Google Scholar] [CrossRef]
  49. Henstra, C.; van Praagh, J.; Olinga, P.; Nagelkerke, A. The gastrointestinal microbiota in colorectal cancer cell migration and invasion. Clin. Exp. Metastasis 2021, 38, 495–510. [Google Scholar] [CrossRef]
  50. Wang, Q.; Yu, C.; Yue, C.; Liu, X. Fusobacterium nucleatum produces cancer stem cell characteristics via EMT-resembling variations. Int. J. Clin. Exp. Pathol. 2020, 13, 1819–1828. [Google Scholar]
  51. Baud, J.; Varon, C.; Chabas, S.; Chambonnier, L.; Darfeuille, F.; Staedel, C. Helicobacter pylori initiates a mesenchymal transition through ZEB1 in gastric epithelial cells. PLoS ONE 2013, 8, e60315. [Google Scholar] [CrossRef] [PubMed]
  52. Marques, M.S.; Melo, J.; Cavadas, B.; Mendes, N.; Pereira, L.; Carneiro, F.; Figueiredo, C.; Leite, M. Afadin Downregulation by Helicobacter pylori Induces Epithelial to Mesenchymal Transition in Gastric Cells. Front. Microbiol. 2018, 9, 2712. [Google Scholar] [CrossRef] [PubMed]
  53. Li, W.T.; Iyangar, A.S.; Reddy, R.; Chakladar, J.; Bhargava, V.; Sakamoto, K.; Ongkeko, W.M.; Rajasekaran, M. The Bladder Microbiome Is Associated with Epithelial-Mesenchymal Transition in Muscle Invasive Urothelial Bladder Carcinoma. Cancers 2021, 13, 3649. [Google Scholar] [CrossRef] [PubMed]
  54. Abd-El-Raouf, R.; Ouf, S.A.; Gabr, M.M.; Zakaria, M.M.; El-Yasergy, K.F.; Ali-El-Dein, B. Escherichia coli foster bladder cancer cell line progression via epithelial mesenchymal transition, stemness and metabolic reprogramming. Sci. Rep. 2020, 10, 18024. [Google Scholar] [CrossRef] [PubMed]
  55. Katz, J.; Onate, M.D.; Pauley, K.M.; Bhattacharyya, I.; Cha, S. Presence of Porphyromonas gingivalis in gingival squamous cell carcinoma. Int. J. Oral. Sci. 2011, 3, 209–215. [Google Scholar] [CrossRef] [PubMed]
  56. Abdulkareem, A.A.; Shelton, R.M.; Landini, G.; Cooper, P.R.; Milward, M.R. Periodontal pathogens promote epithelial-mesenchymal transition in oral squamous carcinoma cells in vitro. Cell Adhes. Migr. 2018, 12, 127–137. [Google Scholar] [CrossRef] [PubMed]
  57. Ha, N.H.; Woo, B.H.; Kim, D.J.; Ha, E.S.; Choi, J.I.; Kim, S.J.; Park, B.S.; Lee, J.H.; Park, H.R. Prolonged and repetitive exposure to Porphyromonas gingivalis increases aggressiveness of oral cancer cells by promoting acquisition of cancer stem cell properties. Tumour Biol. 2015, 36, 9947–9960. [Google Scholar] [CrossRef] [PubMed]
  58. Sztukowska, M.N.; Ojo, A.; Ahmed, S.; Carenbauer, A.L.; Wang, Q.; Shumway, B.; Jenkinson, H.F.; Wang, H.; Darling, D.S.; Lamont, R.J. Porphyromonas gingivalis initiates a mesenchymal-like transition through ZEB1 in gingival epithelial cells. Cell Microbiol. 2016, 18, 844–858. [Google Scholar] [CrossRef]
  59. Peng, Z.; Wan, P.; Deng, Y.; Shen, W.; Liu, R. Lipopolysaccharide exacerbates to the migration, invasion, and epithelial-mesenchymal transition of esophageal cancer cells by TLR4/NF-κB axis. Environ. Toxicol. 2023, 38, 1090–1099. [Google Scholar] [CrossRef]
  60. Nikolaieva, N.; Sevcikova, A.; Omelka, R.; Martiniakova, M.; Mego, M.; Ciernikova, S. Gut Microbiota-MicroRNA Interactions in Intestinal Homeostasis and Cancer Development. Microorganisms 2022, 11, 107. [Google Scholar] [CrossRef]
  61. Zhu, Z.; Huang, J.; Li, X.; Xing, J.; Chen, Q.; Liu, R.; Hua, F.; Qiu, Z.; Song, Y.; Bai, C.; et al. Gut microbiota regulate tumor metastasis via circRNA/miRNA networks. Gut Microbes 2020, 12, 1788891. [Google Scholar] [CrossRef] [PubMed]
  62. Yan, X.; Liu, L.; Li, H.; Qin, H.; Sun, Z. Clinical significance of Fusobacterium nucleatum, epithelial-mesenchymal transition, and cancer stem cell markers in stage III/IV colorectal cancer patients. OncoTargets Ther. 2017, 10, 5031–5046. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, Y.; Weng, W.; Peng, J.; Hong, L.; Yang, L.; Toiyama, Y.; Gao, R.; Liu, M.; Yin, M.; Pan, C.; et al. Fusobacterium nucleatum Increases Proliferation of Colorectal Cancer Cells and Tumor Development in Mice by Activating Toll-Like Receptor 4 Signaling to Nuclear Factor-κB, and Up-regulating Expression of MicroRNA-21. Gastroenterology 2017, 152, 851–866.e824. [Google Scholar] [CrossRef] [PubMed]
  64. Kovacs, T.; Miko, E.; Ujlaki, G.; Yousef, H.; Csontos, V.; Uray, K.; Bai, P. The involvement of oncobiosis and bacterial metabolite signaling in metastasis formation in breast cancer. Cancer Metastasis Rev. 2021, 40, 1223–1249. [Google Scholar] [CrossRef] [PubMed]
  65. Ujlaki, G.; Kovacs, T.; Vida, A.; Kokai, E.; Rauch, B.; Schwarcz, S.; Miko, E.; Janka, E.; Sipos, A.; Hegedus, C.; et al. Identification of Bacterial Metabolites Modulating Breast Cancer Cell Proliferation and Epithelial-Mesenchymal Transition. Molecules 2023, 28, 5898. [Google Scholar] [CrossRef] [PubMed]
  66. Sari, Z.; Miko, E.; Kovacs, T.; Janko, L.; Csonka, T.; Lente, G.; Sebo, E.; Toth, J.; Toth, D.; Arkosy, P.; et al. Indolepropionic Acid, a Metabolite of the Microbiome, Has Cytostatic Properties in Breast Cancer by Activating AHR and PXR Receptors and Inducing Oxidative Stress. Cancers 2020, 12, 2411. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, D.; Cheng, J.; Zhang, J.; Zhou, F.; He, X.; Shi, Y.; Tao, Y. The Role of Respiratory Microbiota in Lung Cancer. Int. J. Biol. Sci. 2021, 17, 3646–3658. [Google Scholar] [CrossRef] [PubMed]
  68. Castro, P.R.; Bittencourt, L.F.F.; Larochelle, S.; Andrade, S.P.; Mackay, C.R.; Slevin, M.; Moulin, V.J.; Barcelos, L.S. GPR43 regulates sodium butyrate-induced angiogenesis and matrix remodeling. Am. J. Physiol. Heart Circ. Physiol. 2021, 320, H1066–H1079. [Google Scholar] [CrossRef]
  69. Nguyen, T.T.; Lian, S.; Ung, T.T.; Xia, Y.; Han, J.Y.; Jung, Y.D. Lithocholic Acid Stimulates IL-8 Expression in Human Colorectal Cancer Cells via Activation of Erk1/2 MAPK and Suppression of STAT3 Activity. J. Cell. Biochem. 2017, 118, 2958–2967. [Google Scholar] [CrossRef]
  70. Saha, S.; Chowdhury, P.; Pal, A.; Chakrabarti, M.K. Downregulation of human colon carcinoma cell (COLO-205) proliferation through PKG-MAP kinase mediated signaling cascade by E. coli heat stable enterotoxin (STa), a potent anti-angiogenic and anti-metastatic molecule. J. Appl. Toxicol. 2008, 28, 475–483. [Google Scholar] [CrossRef]
  71. Yang, S.; Dai, H.; Lu, Y.; Li, R.; Gao, C.; Pan, S. Trimethylamine N-Oxide Promotes Cell Proliferation and Angiogenesis in Colorectal Cancer. J. Immunol. Res. 2022, 2022, 7043856. [Google Scholar] [CrossRef] [PubMed]
  72. Salahshouri, P.; Emadi-Baygi, M.; Jalili, M.; Khan, F.M.; Wolkenhauer, O.; Salehzadeh-Yazdi, A. A Metabolic Model of Intestinal Secretions: The Link between Human Microbiota and Colorectal Cancer Progression. Metabolites 2021, 11, 456. [Google Scholar] [CrossRef] [PubMed]
  73. Wu, J.; Li, H.; Xie, H.; Wu, X.; Lan, P. The malignant role of exosomes in the communication among colorectal cancer cell, macrophage and microbiome. Carcinogenesis 2019, 40, 601–610. [Google Scholar] [CrossRef] [PubMed]
  74. Wong, S.H.; Zhao, L.; Zhang, X.; Nakatsu, G.; Han, J.; Xu, W.; Xiao, X.; Kwong, T.N.Y.; Tsoi, H.; Wu, W.K.K.; et al. Gavage of Fecal Samples from Patients with Colorectal Cancer Promotes Intestinal Carcinogenesis in Germ-Free and Conventional Mice. Gastroenterology 2017, 153, 1621–1633.e1626. [Google Scholar] [CrossRef] [PubMed]
  75. De Spiegeleer, B.; Verbeke, F.; D’Hondt, M.; Hendrix, A.; Van De Wiele, C.; Burvenich, C.; Peremans, K.; De Wever, O.; Bracke, M.; Wynendaele, E. The quorum sensing peptides PhrG, CSP and EDF promote angiogenesis and invasion of breast cancer cells in vitro. PLoS ONE 2015, 10, e0119471. [Google Scholar] [CrossRef] [PubMed]
  76. Wynendaele, E.; Verbeke, F.; D’Hondt, M.; Hendrix, A.; Van De Wiele, C.; Burvenich, C.; Peremans, K.; De Wever, O.; Bracke, M.; De Spiegeleer, B. Crosstalk between the microbiome and cancer cells by quorum sensing peptides. Peptides 2015, 64, 40–48. [Google Scholar] [CrossRef] [PubMed]
  77. Hilmi, M.; Kamal, M.; Vacher, S.; Dupain, C.; Ibadioune, S.; Halladjian, M.; Sablin, M.P.; Marret, G.; Ajgal, Z.C.; Nijnikoff, M.; et al. Intratumoral microbiome is driven by metastatic site and associated with immune histopathological parameters: An ancillary study of the SHIVA clinical trial. Eur. J. Cancer 2023, 183, 152–161. [Google Scholar] [CrossRef] [PubMed]
  78. Hajjar, J.; Mendoza, T.; Zhang, L.; Fu, S.; Piha-Paul, S.A.; Hong, D.S.; Janku, F.; Karp, D.D.; Ballhausen, A.; Gong, J.; et al. Associations between the gut microbiome and fatigue in cancer patients. Sci. Rep. 2021, 11, 5847. [Google Scholar] [CrossRef]
  79. Sethi, V.; Kurtom, S.; Tarique, M.; Lavania, S.; Malchiodi, Z.; Hellmund, L.; Zhang, L.; Sharma, U.; Giri, B.; Garg, B.; et al. Gut Microbiota Promotes Tumor Growth in Mice by Modulating Immune Response. Gastroenterology 2018, 155, 33–37.e36. [Google Scholar] [CrossRef]
  80. Spakowicz, D.; Hoyd, R.; Muniak, M.; Husain, M.; Bassett, J.S.; Wang, L.; Tinoco, G.; Patel, S.H.; Burkart, J.; Miah, A.; et al. Inferring the role of the microbiome on survival in patients treated with immune checkpoint inhibitors: Causal modeling, timing, and classes of concomitant medications. BMC Cancer 2020, 20, 383. [Google Scholar] [CrossRef]
  81. Dzutsev, A.; Goldszmid, R.S.; Viaud, S.; Zitvogel, L.; Trinchieri, G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur. J. Immunol. 2015, 45, 17–31. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, T.; Cai, G.; Qiu, Y.; Fei, N.; Zhang, M.; Pang, X.; Jia, W.; Cai, S.; Zhao, L. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012, 6, 320–329. [Google Scholar] [CrossRef] [PubMed]
  83. Viljoen, K.S.; Dakshinamurthy, A.; Goldberg, P.; Blackburn, J.M. Quantitative profiling of colorectal cancer-associated bacteria reveals associations between Fusobacterium spp., enterotoxigenic Bacteroides fragilis (ETBF) and clinicopathological features of colorectal cancer. PLoS ONE 2015, 10, e0119462. [Google Scholar] [CrossRef] [PubMed]
  84. Xu, C.; Fan, L.; Lin, Y.; Shen, W.; Qi, Y.; Zhang, Y.; Chen, Z.; Wang, L.; Long, Y.; Hou, T.; et al. Fusobacterium nucleatum promotes colorectal cancer metastasis through miR-1322/CCL20 axis and M2 polarization. Gut Microbes 2021, 13, 1980347. [Google Scholar] [CrossRef] [PubMed]
  85. Chen, S.; Su, T.; Zhang, Y.; Lee, A.; He, J.; Ge, Q.; Wang, L.; Si, J.; Zhuo, W.; Wang, L. Fusobacterium nucleatum promotes colorectal cancer metastasis by modulating KRT7-AS/KRT7. Gut Microbes 2020, 11, 511–525. [Google Scholar] [CrossRef] [PubMed]
  86. Chen, S.; Zhang, L.; Li, M.; Zhang, Y.; Sun, M.; Wang, L.; Lin, J.; Cui, Y.; Chen, Q.; Jin, C.; et al. Fusobacterium nucleatum reduces METTL3-mediated m6A modification and contributes to colorectal cancer metastasis. Nat. Commun. 2022, 13, 1248. [Google Scholar] [CrossRef] [PubMed]
  87. Bullman, S.; Pedamallu, C.S.; Sicinska, E.; Clancy, T.E.; Zhang, X.; Cai, D.; Neuberg, D.; Huang, K.; Guevara, F.; Nelson, T.; et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 1443–1448. [Google Scholar] [CrossRef]
  88. Wang, F.; He, M.M.; Yao, Y.C.; Zhao, X.; Wang, Z.Q.; Jin, Y.; Luo, H.Y.; Li, J.B.; Wang, F.H.; Qiu, M.Z.; et al. Regorafenib plus toripalimab in patients with metastatic colorectal cancer: A phase Ib/II clinical trial and gut microbiome analysis. Cell Rep. Med. 2021, 2, 100383. [Google Scholar] [CrossRef]
  89. Ambalam, P.; Raman, M.; Purama, R.K.; Doble, M. Probiotics, prebiotics and colorectal cancer prevention. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 119–131. [Google Scholar] [CrossRef]
  90. Dejea, C.M.; Fathi, P.; Craig, J.M.; Boleij, A.; Taddese, R.; Geis, A.L.; Wu, X.; DeStefano Shields, C.E.; Hechenbleikner, E.M.; Huso, D.L.; et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science 2018, 359, 592–597. [Google Scholar] [CrossRef]
  91. Stakelum, A.; Zaborowski, A.; Collins, D.; Winter, D.C. The influence of the gastrointestinal microbiome on colorectal metastasis: A narrative review. Colorectal Dis. 2020, 22, 1101–1107. [Google Scholar] [CrossRef] [PubMed]
  92. Wachsmannova, L.; Stevurkova, V.; Ciernikova, S. Changes in SNAI1 and VIM gene expression in Caco2 cells after cocultivation with bacteria from colorectal cancer biopsies. Neoplasma 2019, 66, 271–275. [Google Scholar] [CrossRef] [PubMed]
  93. Sun, L.; Yan, Y.; Chen, D.; Yang, Y. Quxie Capsule Modulating Gut Microbiome and Its Association with T cell Regulation in Patients with Metastatic Colorectal Cancer: Result From a Randomized Controlled Clinical Trial. Integr. Cancer Ther. 2020, 19, 1534735420969820. [Google Scholar] [CrossRef] [PubMed]
  94. Li, J.J.; Zhu, M.; Kashyap, P.C.; Chia, N.; Tran, N.H.; McWilliams, R.R.; Bekaii-Saab, T.S.; Ma, W.W. The role of microbiome in pancreatic cancer. Cancer Metastasis Rev. 2021, 40, 777–789. [Google Scholar] [CrossRef] [PubMed]
  95. Jeong, J.Y.; Kim, T.B.; Kim, J.; Choi, H.W.; Kim, E.J.; Yoo, H.J.; Lee, S.; Jun, H.R.; Yoo, W.; Kim, S.; et al. Diversity in the Extracellular Vesicle-Derived Microbiome of Tissues according to Tumor Progression in Pancreatic Cancer. Cancers 2020, 12, 2346. [Google Scholar] [CrossRef] [PubMed]
  96. Ren, Z.; Chen, X.; Hong, L.; Zhao, X.; Cui, G.; Li, A.; Liu, Y.; Zhou, L.; Sun, R.; Shen, S.; et al. Nanoparticle Conjugation of Ginsenoside Rg3 Inhibits Hepatocellular Carcinoma Development and Metastasis. Small 2020, 16, e1905233. [Google Scholar] [CrossRef] [PubMed]
  97. Huang, D.; Su, X.; Yuan, M.; Zhang, S.; He, J.; Deng, Q.; Qiu, W.; Dong, H.; Cai, S. The characterization of lung microbiome in lung cancer patients with different clinicopathology. Am. J. Cancer Res. 2019, 9, 2047–2063. [Google Scholar]
  98. Yu, G.; Gail, M.H.; Consonni, D.; Carugno, M.; Humphrys, M.; Pesatori, A.C.; Caporaso, N.E.; Goedert, J.J.; Ravel, J.; Landi, M.T. Characterizing human lung tissue microbiota and its relationship to epidemiological and clinical features. Genome Biol. 2016, 17, 163. [Google Scholar] [CrossRef]
  99. Gowing, S.D.; Chow, S.C.; Cools-Lartigue, J.J.; Chen, C.B.; Najmeh, S.; Jiang, H.Y.; Bourdeau, F.; Beauchamp, A.; Mancini, U.; Angers, I.; et al. Gram-positive pneumonia augments non-small cell lung cancer metastasis via host toll-like receptor 2 activation. Int. J. Cancer 2017, 141, 561–571. [Google Scholar] [CrossRef]
  100. Jungnickel, C.; Wonnenberg, B.; Karabiber, O.; Wolf, A.; Voss, M.; Wolf, L.; Honecker, A.; Kamyschnikow, A.; Herr, C.; Bals, R.; et al. Cigarette smoke-induced disruption of pulmonary barrier and bacterial translocation drive tumor-associated inflammation and growth. Am. J. Physiol. Lung Cell Mol. Physiol. 2015, 309, L605–L613. [Google Scholar] [CrossRef]
  101. Hadzega, D.; Minarik, G.; Karaba, M.; Kalavska, K.; Benca, J.; Ciernikova, S.; Sedlackova, T.; Nemcova, P.; Bohac, M.; Pindak, D.; et al. Uncovering Microbial Composition in Human Breast Cancer Primary Tumour Tissue Using Transcriptomic RNA-seq. Int. J. Mol. Sci. 2021, 22, 9058. [Google Scholar] [CrossRef] [PubMed]
  102. Buchta Rosean, C.; Bostic, R.R.; Ferey, J.C.M.; Feng, T.Y.; Azar, F.N.; Tung, K.S.; Dozmorov, M.G.; Smirnova, E.; Bos, P.D.; Rutkowski, M.R. Preexisting Commensal Dysbiosis Is a Host-Intrinsic Regulator of Tissue Inflammation and Tumor Cell Dissemination in Hormone Receptor-Positive Breast Cancer. Cancer Res. 2019, 79, 3662–3675. [Google Scholar] [CrossRef] [PubMed]
  103. Wenhui, Y.; Zhongyu, X.; Kai, C.; Zhaopeng, C.; Jinteng, L.; Mengjun, M.; Zepeng, S.; Yunshu, C.; Peng, W.; Yanfeng, W.; et al. Variations in the Gut Microbiota in Breast Cancer Occurrence and Bone Metastasis. Front. Microbiol. 2022, 13, 894283. [Google Scholar] [CrossRef] [PubMed]
  104. Parhi, L.; Alon-Maimon, T.; Sol, A.; Nejman, D.; Shhadeh, A.; Fainsod-Levi, T.; Yajuk, O.; Isaacson, B.; Abed, J.; Maalouf, N.; et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat. Commun. 2020, 11, 3259. [Google Scholar] [CrossRef] [PubMed]
  105. Chiba, A.; Bawaneh, A.; Velazquez, C.; Clear, K.Y.J.; Wilson, A.S.; Howard-McNatt, M.; Levine, E.A.; Levi-Polyachenko, N.; Yates-Alston, S.A.; Diggle, S.P.; et al. Neoadjuvant Chemotherapy Shifts Breast Tumor Microbiota Populations to Regulate Drug Responsiveness and the Development of Metastasis. Mol. Cancer Res. 2020, 18, 130–139. [Google Scholar] [CrossRef] [PubMed]
  106. Eun, Y.G.; Lee, J.W.; Kim, S.W.; Hyun, D.W.; Bae, J.W.; Lee, Y.C. Oral microbiome associated with lymph node metastasis in oral squamous cell carcinoma. Sci. Rep. 2021, 11, 23176. [Google Scholar] [CrossRef] [PubMed]
  107. Qiao, H.; Tan, X.R.; Li, H.; Li, J.Y.; Chen, X.Z.; Li, Y.Q.; Li, W.F.; Tang, L.L.; Zhou, G.Q.; Zhang, Y.; et al. Association of Intratumoral Microbiota with Prognosis in Patients with Nasopharyngeal Carcinoma from 2 Hospitals in China. JAMA Oncol. 2022, 8, 1301–1309. [Google Scholar] [CrossRef]
  108. Yuan, L.; Yang, P.; Wei, G.; Hu, X.; Chen, S.; Lu, J.; Yang, L.; He, X.; Bao, G. Tumor microbiome diversity influences papillary thyroid cancer invasion. Commun. Biol. 2022, 5, 864. [Google Scholar] [CrossRef]
  109. Peiffer, L.B.; White, J.R.; Jones, C.B.; Slottke, R.E.; Ernst, S.E.; Moran, A.E.; Graff, J.N.; Sfanos, K.S. Composition of gastrointestinal microbiota in association with treatment response in individuals with metastatic castrate resistant prostate cancer progressing on enzalutamide and initiating treatment with anti-PD-1 (pembrolizumab). Neoplasia 2022, 32, 100822. [Google Scholar] [CrossRef]
  110. Ma, J.; Gnanasekar, A.; Lee, A.; Li, W.T.; Haas, M.; Wang-Rodriguez, J.; Chang, E.Y.; Rajasekaran, M.; Ongkeko, W.M. Influence of Intratumor Microbiome on Clinical Outcome and Immune Processes in Prostate Cancer. Cancers 2020, 12, 2524. [Google Scholar] [CrossRef]
  111. Salgia, N.J.; Bergerot, P.G.; Maia, M.C.; Dizman, N.; Hsu, J.; Gillece, J.D.; Folkerts, M.; Reining, L.; Trent, J.; Highlander, S.K.; et al. Stool Microbiome Profiling of Patients with Metastatic Renal Cell Carcinoma Receiving Anti-PD-1 Immune Checkpoint Inhibitors. Eur. Urol. 2020, 78, 498–502. [Google Scholar] [CrossRef]
  112. Agarwal, A.; Modliszewski, J.; Davey, L.; Reyes-Martinez, M.; Runyambo, D.; Corcoran, D.; Dressman, H.; George, D.J.; Valdivia, R.H.; Armstrong, A.J.; et al. Investigating the role of the gastrointestinal microbiome in response to immune checkpoint inhibitors (ICIs) among patients (pts) with metastatic renal cell carcinoma (mRCC). J. Clin. Oncol. 2020, 38, 730. [Google Scholar] [CrossRef]
  113. Dizman, N.; Hsu, J.; Bergerot, P.G.; Gillece, J.D.; Folkerts, M.; Reining, L.; Trent, J.; Highlander, S.K.; Pal, S.K. Randomized trial assessing impact of probiotic supplementation on gut microbiome and clinical outcome from targeted therapy in metastatic renal cell carcinoma. Cancer Med. 2021, 10, 79–86. [Google Scholar] [CrossRef] [PubMed]
  114. Hahn, A.W.; Froerer, C.; VanAlstine, S.; Rathi, N.; Bailey, E.B.; Stenehjem, D.D.; Agarwal, N. Targeting Bacteroides in Stool Microbiome and Response to Treatment with First-Line VEGF Tyrosine Kinase Inhibitors in Metastatic Renal-Cell Carcinoma. Clin. Genitourin. Cancer 2018, 16, 365–368. [Google Scholar] [CrossRef] [PubMed]
  115. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [PubMed]
  116. Mizuhashi, S.; Kajihara, I.; Sawamura, S.; Kanemaru, H.; Makino, K.; Aoi, J.; Makino, T.; Masuguchi, S.; Fukushima, S.; Ihn, H. Skin microbiome in acral melanoma: Corynebacterium is associated with advanced melanoma. J. Dermatol. 2021, 48, e15–e16. [Google Scholar] [CrossRef] [PubMed]
  117. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [PubMed]
  118. Wargo, J.A.; Gopalakrishnan, V.; Spencer, C.; Karpinets, T.; Reuben, A.; Andrews, M.C.; Tetzlaff, M.T.; Lazar, A.; Hwu, P.; Hwu, W.J.; et al. Association of the diversity and composition of the gut microbiome with responses and survival (PFS) in metastatic melanoma (MM) patients (pts) on anti-PD-1 therapy. J. Clin. Oncol. 2017, 15, 3008. [Google Scholar] [CrossRef]
  119. Vitali, F.; Colucci, R.; Di Paola, M.; Pindo, M.; De Filippo, C.; Moretti, S.; Cavalieri, D. Early melanoma invasivity correlates with gut fungal and bacterial profiles. Br. J. Dermatol. 2022, 186, 106–116. [Google Scholar] [CrossRef]
  120. Ciernikova, S.; Mego, M.; Hainova, K.; Adamcikova, Z.; Stevurkova, V.; Zajac, V. Modification of microflora imbalance: Future directions for prevention and treatment of colorectal cancer? Neoplasma 2015, 62, 345–352. [Google Scholar] [CrossRef]
  121. Wang, Z.; Li, L.; Wang, S.; Wei, J.; Qu, L.; Pan, L.; Xu, K. The role of the gut microbiota and probiotics associated with microbial metabolisms in cancer prevention and therapy. Front. Pharmacol. 2022, 13, 1025860. [Google Scholar] [CrossRef] [PubMed]
  122. Luo, M.; Hu, M.; Xu, F.; Wu, X.; Dong, D.; Wang, W. Preventive effect of Lactobacillus reuteri on melanoma. Biomed. Pharmacother. 2020, 126, 109929. [Google Scholar] [CrossRef] [PubMed]
  123. Pereira, F.V.; Melo, A.C.L.; Silva, M.B.; de Melo, F.M.; Terra, F.F.; Castro, I.A.; Perandini, L.A.; Miyagi, M.T.; Sato, F.T.; Origassa, C.S.T.; et al. Interleukin-6 and the Gut Microbiota Influence Melanoma Progression in Obese Mice. Nutr. Cancer 2021, 73, 642–651. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, L.; Zhou, X.; Wang, Y.; Wang, D.; Ke, Y.; Zeng, X. Propionate and Butyrate Produced by Gut Microbiota after Probiotic Supplementation Attenuate Lung Metastasis of Melanoma Cells in Mice. Mol. Nutr. Food Res. 2021, 65, e2100096. [Google Scholar] [CrossRef] [PubMed]
  125. Le Noci, V.; Guglielmetti, S.; Arioli, S.; Camisaschi, C.; Bianchi, F.; Sommariva, M.; Storti, C.; Triulzi, T.; Castelli, C.; Balsari, A.; et al. Modulation of Pulmonary Microbiota by Antibiotic or Probiotic Aerosol Therapy: A Strategy to Promote Immunosurveillance against Lung Metastases. Cell Rep. 2018, 24, 3528–3538. [Google Scholar] [CrossRef] [PubMed]
  126. Li, J.; Sung, C.Y.; Lee, N.; Ni, Y.; Pihlajamaki, J.; Panagiotou, G.; El-Nezami, H. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc. Natl. Acad. Sci. USA 2016, 113, E1306–E1315. [Google Scholar] [CrossRef] [PubMed]
  127. Dizman, N.; Meza, L.; Bergerot, P.; Alcantara, M.; Dorff, T.; Lyou, Y.; Frankel, P.; Cui, Y.; Mira, V.; Llamas, M.; et al. Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: A randomized phase 1 trial. Nat. Med. 2022, 28, 704–712. [Google Scholar] [CrossRef]
  128. Jakubauskas, M.; Jakubauskiene, L.; Leber, B.; Horvath, A.; Strupas, K.; Stiegler, P.; Schemmer, P. Probiotic Supplementation Attenuates Chemotherapy-Induced Intestinal Mucositis in an Experimental Colorectal Cancer Liver Metastasis Rat Model. Nutrients 2023, 15, 1117. [Google Scholar] [CrossRef]
  129. Shang, F.; Jiang, X.; Wang, H.; Chen, S.; Wang, X.; Liu, Y.; Guo, S.; Li, D.; Yu, W.; Zhao, Z.; et al. The inhibitory effects of probiotics on colon cancer cells: In vitro and in vivo studies. J. Gastrointest. Oncol. 2020, 11, 1224–1232. [Google Scholar] [CrossRef]
  130. Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021, 371, 602–609. [Google Scholar] [CrossRef]
Figure 1. The key processes involved in tumor progression and metastasis. A deep understanding of the crucial events and corresponding mechanisms leading to the formation of distant metastases is essential for developing treatment modalities to target different stages of cancer development and improve patient outcomes.
Figure 1. The key processes involved in tumor progression and metastasis. A deep understanding of the crucial events and corresponding mechanisms leading to the formation of distant metastases is essential for developing treatment modalities to target different stages of cancer development and improve patient outcomes.
Ijms 24 17199 g001
Figure 2. The involvement of the gut and intratumoral microbiome in metastatic processes. Not only cancer development but also the type of anti-cancer therapy affects the diversity of microbial composition in the gastrointestinal tract and alters microbial-associated metabolites. The deregulated thickness of the gut mucosal layer might be responsible for bacterial translocation and development of bloodstream infections. Gastrointestinal dysbiosis results in the inflammation that promotes cancer cell spread due to changed immune responses. Microbes within the tumor microenvironment affect the progression of cancer via modulated immunity and changed inflammatory signaling pathways. Moreover, studies observed the relationship between intratumoral bacteria and metastasis via increased resistance to mechanical stress. Abbreviations: EMT, epithelial-to-mesenchymal transition.
Figure 2. The involvement of the gut and intratumoral microbiome in metastatic processes. Not only cancer development but also the type of anti-cancer therapy affects the diversity of microbial composition in the gastrointestinal tract and alters microbial-associated metabolites. The deregulated thickness of the gut mucosal layer might be responsible for bacterial translocation and development of bloodstream infections. Gastrointestinal dysbiosis results in the inflammation that promotes cancer cell spread due to changed immune responses. Microbes within the tumor microenvironment affect the progression of cancer via modulated immunity and changed inflammatory signaling pathways. Moreover, studies observed the relationship between intratumoral bacteria and metastasis via increased resistance to mechanical stress. Abbreviations: EMT, epithelial-to-mesenchymal transition.
Ijms 24 17199 g002
Table 1. Exploring the microbial markers associated with treatment outcomes in advanced or metastatic cancer patients (according to https://ClinicalTrials.gov/, accessed on 18 October 2023).
Table 1. Exploring the microbial markers associated with treatment outcomes in advanced or metastatic cancer patients (according to https://ClinicalTrials.gov/, accessed on 18 October 2023).
StudyStudy DesignDiseasePurposePatients (n)InterventionStudy Status
NCT03941080An observational prospective studyMetastatic CRCTo confirm the microbial taxa associated with treatment response and side effects in metastatic or irresectable disease300 adults/
older adults
Enrolled patients will be newly diagnosed with an indication for standard palliative systemic treatment.Recruiting
NCT04579484An observational prospective studyMetastatic breast cancerTo determine the gut microbiome in fecal samples of patients with ER+ HER2 breast cancer and assess the relationship between dietary factors and microbiome20 adults/
older adults
Patients will receive endocrine therapy with an aromatase inhibitor combined with an inhibitor of cyclin-dependent kinases 4 and 6.Recruiting
NCT04804956An observational prospective studyMetastatic rectal cancerTo identify the profile of the mesorectal microbiome and correlation with poor prognosis prediction100 adultsParticipants will receive neoadjuvant treatment.Recruiting
NCT04579978An observational prospective studyMetastatic solid cancerTo study changes in the gut microbial community after ICI and evaluate bacterial species associated with treatment efficacy60 adults/
older adults
Patients will be enrolled in the study for planned standard-of-care ICI.Recruiting
NCT05878977An interventional open-label studyMetastatic melanomaTo define novel markers for the prediction of therapy response150 adults/
older adults
Immunotherapy will consist of PD-1 and CTLA-4 inhibitors.Recruiting
NCT05635149An observational prospective studyMetastatic CRCTo assess the composition of the gut microbiome and its association with treatment efficacy100 adults/
older adults
Patients will be treated with Fruquintinib, ICI plus RT, or Fruquintinib and ICI alone.Recruiting
NCT05753839An interventional randomized open-label study with parallel assignmentMetastatic clear cell renal cell carcinoma/kidney cancerTo correlate the gut and urine microbiome compositions with OS, PFS, and ORR40 adults/
older adults
Patients will receive ICI followed by maintenance therapy with ICI or cytoreductive nephrectomy ± metastasectomy after ICI.Recruiting
NCT04090710An interventional randomized study with parallel assignmentMetastatic renal cell carcinomaTo investigate the changes in the gut microbiome via analysis of stool samples78 children/
adults/
older adults
Patients will undergo cytoreductive stereotactic body RT with a combination of ICIs vs. one ICI alone.Recruiting
NCT04243720An observational prospective studyMetastatic solid cancerTo determine changes in the gut microbiome associated with resistance to immunotherapy100 adults/
older adults
Only participants who progressed on immunotherapy will be enrolled in this study.Recruiting
NCT04148378An observational case-only prospective studyCRC neoplasms/
metastatic CRC/
colorectal sarcoma/
adenocarcinoma
To correlate microbiome composition with type of disease100 children/
adults/
older adults
There is no intervention for the study.Unknown
NCT04516135An interventional randomized open-label study with parallel assignmentMetastatic gynecologic cancersTo describe overall diversity, richness, and specific microbial dynamics in the gut and vaginal microbiomes 108 adults/
older adults
Females will be treated with 3D conformal RT/intensity-modulated RT/volume-modulated arc therapy at the physician’s discretion for 1 fraction in the absence of RT-induced toxicities or progression.Recruiting
NCT04214015An observational case-only prospective studyMetastatic mesotheliomaTo analyze the relative abundance of bacterial members in the gut microbiome100 children/
adults/
older adults
There is no intervention for the study.Unknown
NCT03818061An interventional non-randomized study with parallel assignmentMetastatic HNSCCTo characterize the gut microbiome in immunotherapy using whole-metagenome sequencing 33 adults/
older adults
Patients with/without human papillomavirus will receive atezolizumab combined with bevacizumab.Active, but not recruiting
NCT03698461An interventional open-label study with single-group assignmentMetastatic neoplasms/
colorectal neoplasms/
colonic neoplasms/
rectal neoplasms
To determine fecal microbial profile in different time frames20 adults/
older adults
Anti-cancer treatment will consist of atezolizumab with bevacizumab, levoleucovorin, oxaliplatin, and 5-fluorouracil.Active, but not recruiting
NCT03977571An interventional randomized open-label study with parallel assignmentMetastatic renal cell carcinoma/
kidney cancer/
synchronous neoplasm
To correlate the gut microbiome with OS, PFS, and ORR400 adults/
older adults
Patients will receive deferred cytoreductive nephrectomy/no surgery following nivolumab with ipilimumab or tyrosine kinase inhibitors.Recruiting
NCT04636775An observational prospective studyMetastatic non-small-cell lung cancerTo assess the correlations between gut microbiome composition and adverse effects and differences between responders and non-responders46 adults/
older adults
Patients will be treated with immunotherapy using ICI.Recruiting
NCT04219137An observational prospective studyMetastatic EGATo study the microbiome in feces and rectal swab samples120 adults/
older adults
Participants will undergo platinum-based chemotherapy.Unknown
NCT03161756An interventional non-randomized study parallel assignmentMetastatic melanomaTo explore associations between the gut microbiome and therapy response72 adults/
older adults
Nivolumab alone or in combination with ipilimumab will be administered intravenously plus denosumab subcutaneously.Active, but not recruiting
NCT04720768An interventional open-label study with sequential assignmentMetastatic melanomaTo identify fecal biomarkers associated with therapy response/resistance78 adults/
older adults
Patients will receive combined treatment with encorafenib, binimetinib, and palbociclib.Recruiting
NCT03340129An interventional randomized open-label study with parallel assignmentMetastatic melanomaTo observe the diversity and composition of the gut microbiome and to determine the correlation between mucosal integrity and microbes218 adults/
older adults
Treatment will include ipilimumab and nivolumab with concurrent intracranial stereotactic RT or ipilimumab and nivolumab alone.Recruiting
Abbreviations: CRC, colorectal cancer; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; EGA, esophagogastric adenocarcinoma; HNSCC, head and neck squamous cell carcinoma; ICI, immune checkpoint inhibitors; ORR, overall response rate; OS, overall survival; PD-1, programed cell death protein 1; PFS, progression-free survival; RT, radiotherapy.
Table 2. Detection of specific microorganisms in advanced/metastatic cancer. The table summarizes fundamental preclinical/clinical studies and their major findings.
Table 2. Detection of specific microorganisms in advanced/metastatic cancer. The table summarizes fundamental preclinical/clinical studies and their major findings.
MalignancyStudy Type Preclinical/ClinicalInterventionChanges in Microbial CompositionMajor FindingsRef.
CRC with liver/lung metastasesPatientsRegorafenib plus toripalimabFusobacterium, Alistipes, Bilophila, and AcidaminococcusA higher level of specific bacteria was observed in non-responders. Shorter PFS correlated with a higher amount of Fusobacterium.[88]
FAPPatients/
mice
No intervention providedE. coli and ETBFBoth bacterial taxa were biofilm members in FAP tissues from patients. Colonization with E. coli and ETBF increased DNA damage and IL-17 production in carcinogen-treated mice.[90]
CRC with liver/lung metastasesPatientsQuxie capsulesActinobacteria, Oscillibacter, Eubacterium, and LachnospiraceaeCapsules increased butyrate-producing, immunity-stimulating, and anti-cancer bacterial taxa and enhanced Th cells, both CD4 and CD8 cells.[93]
PDAC with lymph node metastasesPatientsNo interventionLeuconostoc, Sutterella, Comamonas, and TuricibacterLower levels of Leuconostoc and Sutterella were documented in tumors with a size ≥3 cm. An increase in lymph node metastases correlated with a higher abundance of Comamonas and Turicibacter. On the contrary, Streptococcus dominated recurrence-free tumors.[95]
Hepatocellular carcinomaMiceNpRg3Bacteroidetes, Verrucomicrobia, and FirmicutesDeveloped NpRg3 remodeled gut microbiome via reduced Firmicutes and increased Bacteroidetes and Verrucomicrobia in stool samples. Moreover, NpRg3 attenuated tumor development and lung metastatic formation in dimethylnitrosamine-induced spontaneous murine carcinoma.[96]
Lung cancerPatientsSystemic therapy/surgical resectionLegionella and ThermusThermus was abundant in the lung microbiome in patients with advanced cancer stages, while Legionella was enriched in patients with developed metastases. Alpha diversity in tumor tissues was lower than in non-malignant lung tissue samples.[98]
Hormone receptor-positive breast cancerMiceAntibiotic cocktail (vancomycin, ampicillin, metronidazole, neomycin, and gentamicin)Blautia, Alistipes, Blautia, Escherichia/Shigella, and BilophilaOrally gavaged antibiotics caused commensal dysbiosis with a higher abundance of specific genera in poorly metastatic mice. Antibiotics promoted tumor cell dissemination to the lungs/peripheral blood/and lymph nodes.[102]
Breast cancerPatientsNo interventionStreptococcus, Campylobacter, Moraxellaceae, Lactobacillales, Bacilli, Epsilonproteobacteria, Veillonella, Acinetobacter, Pseudomonadales, Megamonas, and AkkermansiaListed bacteria, except for Megamonas and Akkermansia, were increased in stool samples of patients with bone metastases. However, the results showed lowered levels of Megamonas and Akkermansia. Bacterial diversity was reduced in the order of normal controls, patients without metastases, and patients with bone metastases.[103]
Breast cancerPatientsNeoadjuvant chemotherapyStreptococcus, Pseudomonas, Brevundimonas, and StaphylococcusChemotherapy decreased intratumoral Streptococcus and increased Pseudomonas. The development of distant metastases correlated with a higher presence of Brevundimonas and Staphylococcus in primary breast tumors.[105]
Oral squamous cell carcinomaPatientsTherapeutic neck dissection due to positive lymph node metastasesTannerella, Fusobacterium, Prevotella, Stomatobaculum, Bifidobacterium, Finegoldia Peptostreptococcaceae, and ShuttleworthiaTwo taxa—Tannerella and Fusobacterium—were enriched in the oral microbiome of patients without metastases. Other genera from the listed panel increased in patients with developed lymph node metastases. Differences in alpha diversity between the oral microbiome of 2 analyzed groups were not significant.[106]
Castrate-resistant prostate cancerPatientsImmune checkpoint inhibitor (pembrolizumab)A. muciniphila, B. thetaiotaomicron, B. fragilis, and R. unassignedA. muciniphila was depleted in pembrolizumab responders, while other listed microbes were higher in responding patients.[109]
Renal cell carcinomaPatientsImmune checkpoint inhibitor (nivolumab or nivolumab plus ipilimumabA. muciniphila, B. adolescentis,
B. intestinihominis, Odoribacter splanchnicus, Bacteroides ovatus, and Eggerthella lenta
A. muciniphila, B. adolescentis, B. intestinihominis, and O. splanchnicus correlated with clinical benefit in metastatic patients, while B. ovatus and E. lenta were associated with no clinical benefit from immunotherapy.[111]
Renal cell carcinomaPatientsImmune checkpoint inhibitorAkkermansiaThe presence of Akkermansia was documented in both responding and non-responding patients to immunotherapy. Therefore, host-specific or tumor factors might affect therapy response.[112]
MelanomaPatientsImmune checkpoint inhibitorLactobacillales, Clostridiales/Ruminococcaceae, Faecalibacterium, Bacteroidales, B. thetaiotaomicron, E. coli, and Anaerotruncus colihominisLactobacillales dominated the oral microbiome of all metastatic patients. Clostridiales/Ruminococcaceae, Faecalibacterium, and alpha diversity were greater in responders, while Bacteroidales, B. thetaiotaomicron, E. coli, and A. colihominis were abundant in non-responders.[115]
MelanomaPatientsImmune checkpoint inhibitorB. longum, C. aerofaciens, E. faecium, R. obeum, and R. intestinalisThe authors observed a higher abundance of R. obeum and R. intestinalis in non-responders, while the other 3 species were enriched significantly in the responder gut microbiome.[117]
MelanomaPatientsImmune checkpoint inhibitorClostridiales and BacteroidalesHigher bacterial diversity with the prevalence of Clostridiales was observed in the gut microbiome of responding patients. However, the dominance of Bacteroidales within the gut microbiome characterized non-responders.[118]
MelanomaPatientsNo interventionCorynebacteriumIn swab samples, Corynebacterium was the most detected taxa in advanced-stage patients. However, the authors did not detect associations between cutaneous microbiome and cancer stage.[116]
Abbreviations: ETBF, enterotoxigenic B. fragilis; FAP, familial adenomatous polyposis; NpRg3, nanoparticle conjugation of ginsenoside Rg3; PDAC, pancreatic ductal adenocarcinoma; PFS, progression-free survival; Th cells, T helper cells.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sevcikova, A.; Mladosievicova, B.; Mego, M.; Ciernikova, S. Exploring the Role of the Gut and Intratumoral Microbiomes in Tumor Progression and Metastasis. Int. J. Mol. Sci. 2023, 24, 17199. https://doi.org/10.3390/ijms242417199

AMA Style

Sevcikova A, Mladosievicova B, Mego M, Ciernikova S. Exploring the Role of the Gut and Intratumoral Microbiomes in Tumor Progression and Metastasis. International Journal of Molecular Sciences. 2023; 24(24):17199. https://doi.org/10.3390/ijms242417199

Chicago/Turabian Style

Sevcikova, Aneta, Beata Mladosievicova, Michal Mego, and Sona Ciernikova. 2023. "Exploring the Role of the Gut and Intratumoral Microbiomes in Tumor Progression and Metastasis" International Journal of Molecular Sciences 24, no. 24: 17199. https://doi.org/10.3390/ijms242417199

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop